Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Mol Med Rep ; 20(5): 4067-4072, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31545433

ABSTRACT

Brucellosis, caused by a facultative intracellular parasite Brucella species, is the most common bacterial zoonotic infection worldwide. Brucella can survive and proliferate in several phagocytic and non­phagocytic cell types. Human brucellosis has similar clinical symptoms with systemic diseases, which may lead to delay of diagnosis and increasing of complications. Therefore, investigating the proliferation of Brucella in host cells is important to understand the pathogenesis of the disease. Dihydroartemisinin (DHA), a semi­synthetic derivative of artemisinin, has been recommended by World Health Organization as an anti­malarial drug. However, there have been few studies regarding its effectiveness against bacteria. In the present study, it was revealed that B. suis vaccine strain 2 (S2) grew in BV2 cells without significant cytotoxicity, and less than 20 µM DHA had no inhibitory effects on BV2 cells. Furthermore, DHA reduced B. suis S2 growth in BV2 cells, and increased the percentage of apoptosis and the expression of cleaved caspase­3 in B. suis S2­infected cells. Collectively, the present data indicated that DHA induced the caspase­dependent apoptotic pathway to inhibit the intracellular B. suis S2 growth.


Subject(s)
Apoptosis/drug effects , Artemisinins/pharmacology , Brucella suis/drug effects , Brucella suis/physiology , Microbial Viability/drug effects , Microglia/microbiology , Animals , Brucellosis/metabolism , Brucellosis/microbiology , Humans , Mice
2.
BMC Vet Res ; 15(1): 304, 2019 Aug 22.
Article in English | MEDLINE | ID: mdl-31438945

ABSTRACT

BACKGROUND: Brucellosis is a worldwide zoonotic infectious disease that is transmitted in various ways and causes great harm to humans and animals. The brucellosis pathogen is Brucella, which mainly resides in macrophage cells and survives and replicates in host cells. However, the mechanisms underlying Brucella survival in macrophage cells have not been thoroughly elucidated to date. Peroxiredoxin 6 (Prdx6) is a bifunctional protein that shows not only GSH peroxidase activity but also phospholipase A2 activity and plays important roles in combating oxidative damage and regulating apoptosis. RESULTS: Recombinant mouse (Mus musculus) Prdx6 (MmPrdx6) was expressed and purified, and monoclonal antibodies against MmPrdx6 were prepared. Using the Brucella suis S2 strain to infect RAW264.7 murine macrophages, the level of intracellular Prdx6 expression first decreased and later increased following infection. Overexpressing Prdx6 in macrophages resulted in an increase in B. suis S2 strain levels in RAW264.7 cells, while knocking down Prdx6 reduced the S2 levels in cells. CONCLUSIONS: Host Prdx6 can increase the intracellular survival of B. suis S2 strain and plays a role in Brucella infection.


Subject(s)
Brucella suis/physiology , Brucellosis/microbiology , Peroxiredoxin VI/metabolism , Animals , Female , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Macrophages/metabolism , Mice , Mice, Inbred BALB C , RAW 264.7 Cells
3.
Front Immunol ; 10: 1775, 2019.
Article in English | MEDLINE | ID: mdl-31402921

ABSTRACT

Brucella enters their hosts mostly through mucosae from where it spreads systemically. Adhesion to extracellular matrix (ECM) components or to host cells is important for the infectious process, and is mediated by several adhesins, including the BtaF trimeric autotransporter. Although Th1 responses and gamma interferon (IFN-γ) are important for protection, antibodies able to block adhesions might also contribute to prevent Brucella infection. We evaluated the importance of BtaF for respiratory Brucella infection, and characterized the immune response and protection from mucosal challenge induced by nasal vaccination with recombinant BtaF. While lung CFU numbers did not differ at day 1 p.i. between mice intratracheally inoculated with B. suis M1330 (wild type) and those receiving a ΔbtaF mutant, they were reduced in the latter group at 7 and 30 days p.i. For vaccination studies the BtaF passenger domain was engineered and expressed as a soluble trimeric protein. Mice were immunized by the nasal route with BtaF or saline (control group) plus the mucosal adjuvant c-di-AMP. Specific anti-BtaF antibodies (IgG and IgA) were increased in serum, including a mixed IgG2a/IgG1 response. In vitro, these antibodies reduced bacterial adhesion to A549 alveolar epithelial cells. Specific IgA antibodies were also increased in several mucosae. Spleen cells from BtaF immunized mice significantly increased their IL-2, IL-5, IL-17, and IFN-γ secretion upon antigen stimulation. In cervical draining lymph nodes, antigen-experienced CD4+ T cells were maintained mainly as central memory cells. A BtaF-specific delayed-type hypersensitivity response was detected in BtaF immunized mice. Lung cells from the latter produced high levels of IFN-γ upon antigen stimulation. Although nasal immunization with BtaF did not protect mice against B. suis respiratory challenge, it conferred significant protection from intragastric challenge; the splenic load of B. suis was reduced by 3.28 log CFU in immunized mice. This study shows that nasal vaccination with BtaF+c-di-AMP protects against intragastric challenge with B. suis by inducing local and systemic antibody responses, central memory CD4+ T cells and strong Th1 responses. Therefore, although BtaF vaccination did not protect from B. suis respiratory infection, this adhesin constitutes a promising immunogen against mucosal B. suis infection.


Subject(s)
Adhesins, Bacterial/genetics , Antigens, Bacterial/immunology , Brucella suis/physiology , Brucellosis/immunology , Brucellosis/microbiology , Adaptive Immunity , Adhesins, Bacterial/metabolism , Administration, Intranasal , Animals , CD4-Positive T-Lymphocytes , Dinucleoside Phosphates/metabolism , Female , Humans , Immunity, Mucosal/immunology , Immunization/methods , Mice , Virulence
4.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 31(7): 914-7, 2015 Jul.
Article in Chinese | MEDLINE | ID: mdl-26146061

ABSTRACT

OBJECTIVE: To investigate the role of the dead macrophages infected by Brucella suis S2 strain in the initiation of immune response to Brucella. METHODS: The mouse peritoneal macrophages were infected with Brucella suis S2 strain in vitro. After one hour, the cells were cultured in serum-free RPMI1640 medium for 5 days until all of them were dead because of starvation. The dead cell fragments and the bone marrow-derived dendritic cells (BMDCs) were co-cultured for 24, 48 and 72 hours, and then interleukin 12 (IL-12), tumor necrosis factor α (TNF-α) in the co-cultivation supernatant were detected by ELISA. The mouse macrophages marked by carboxyfluorescein diacetate succinimidyl ester (CFSE) were infected with Brucella suis S2 strain in vitro, and then were cultured without serum in the dark; the dead macrophages fragments and BMDCs labeled with anti-CD11c-PE were co-cultured for 1 hour away from light, and then the changes that BMDCs swallowed the fragments of macrophages were observed by laser scanning confocal microscopy. BALB/c mice were inoculated with the fragments of dead macrophages infected by S2 strain through abdominal cavity. After one week, a second immunization was done. The serum levels of IL-4, IL-2 and IFN-γ were detected with ELISA at 3 days post-secondary immunization. RESULTS: The macrophages fragments infected by S2 strain could be swallowed by DCs. The level of TNF-α in BMDCs swallowing macrophages fragments infected by S2 strain was significantly higher than that in the control group, but the former did not secret IL-12. The levels of IL-2, IL-4 and IFN-γ in the sera from the mice inoculated with the macrophages fragments infected by S2 strain were dramatically higher than those in the control groups. CONCLUSION: The dead macrophages infected by Brucella suis S2 strain can activate DCs to present antigen and induce the anti-Brucella immune response.


Subject(s)
Brucella suis/physiology , Brucellosis/immunology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/microbiology , Animals , Brucella suis/immunology , Brucellosis/blood , Cell Death , Cell Movement/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Interferon-gamma/blood , Interleukin-2/blood , Interleukin-4/blood , Lymphocyte Activation/immunology , Macrophages, Peritoneal/immunology , Male , Mice , Tumor Necrosis Factor-alpha/metabolism
5.
Methods Mol Biol ; 1197: 3-9, 2014.
Article in English | MEDLINE | ID: mdl-25172272

ABSTRACT

The use of animal models is a key step to better understand bacterial virulence factors and their roles in host/pathogen interactions. To avoid the ethical and cost problems of mammalian models in bacterial virulence research, several insect models have been developed. One of these models, the larvae of the greater wax moth Galleria mellonella, has been shown to be relevant for several fungal and bacterial mammalian pathogens. Here, we describe the use G. mellonella to study virulence of the highly virulent facultative intracellular bacterial pathogens: Brucella suis, Brucella melitensis, Francisella tularensis, Burkholderia mallei, and Burkholderia pseudomallei.


Subject(s)
Disease Models, Animal , Moths , Animals , Bacterial Infections/microbiology , Brucella melitensis , Brucella suis/physiology , Burkholderia mallei , Burkholderia pseudomallei/physiology , Francisella tularensis/physiology
6.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 29(11): 1137-40, 2013 Nov.
Article in Chinese | MEDLINE | ID: mdl-24200058

ABSTRACT

OBJECTIVE: To study the pattern recognition and activation effect of mast cells infected by Brucella (B.) suis S2. METHODS: Mast cells derived from bone marrow in vitro were infected by B.suis S2. The change in the cell morphology was observed with Wrights-Giemsa's staining, and cell degranulation was tested with toluidine blue staining. The extracellular levels of histamine, IFN-γ and IL-12 of mast cells at 1 and 12 h after infection were detected by indirect ELISA. The uptake pattern of mast cells to B.suis S2 was determined by laser-scanning confocal microscopy. The expressions of TLR4 and TLR8 mRNA were detected by RT-PCR at 12 and 24 h after infection by B.suis S2, and the TLR4 and TLR8 protein expressions were detected by flow cytometry at 24 h. RESULTS: The form of mast cells infected by B.suis S2 was obviously changed. Significant degranulation was observed at 1 h, and at 1, 12 h post-infection by B.suis S2, the content of histamine secreted by mast cells was significantly higher than that of normal control group (P<0.05), and IFN-γ and IL-12 were not found. At 30 min and 1 h, B.suis S2 bound to the mast cell surface and were not uptaken into the mast cells. Compared with the control group, the expression of TLR4 mRNA increased after 12 h infection by B.suis S2, and was reduced at 24 h. The expression of TLR4 protein rose at 24 h, but the expression of TLR8 mRNA and protein did not alter at 12 and 24 h after infection by B.suis S2. CONCLUSION: B.suis S2 can bind to the cell surface and activate mast cells, cause their degranulation, induce the release of histamine, but IFN-γ and IL-12 were not found during the observing time. The mechanism may be that B.suis S2 can be recognized by mast cells through TLR4 but can not be phagocytosed by mast cells.


Subject(s)
Brucella suis/physiology , Mast Cells/cytology , Mast Cells/microbiology , Animals , Bone Marrow Cells/cytology , Cell Degranulation , Female , Male , Mast Cells/metabolism , Mice, Inbred BALB C , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 8/metabolism
7.
PLoS One ; 8(11): e79770, 2013.
Article in English | MEDLINE | ID: mdl-24236157

ABSTRACT

The adhesion of bacterial pathogens to host cells is an event that determines infection, and ultimately invasion and intracellular multiplication. Several evidences have recently shown that this rule is also truth for the intracellular pathogen Brucella. Brucella suis displays the unipolar BmaC and BtaE adhesins, which belong to the monomeric and trimeric autotransporter (TA) families, respectively. It was previously shown that these adhesins are involved in bacterial adhesion to host cells and components of the extracellular matrix (ECM). In this work we describe the role of a new member of the TA family of B. suis (named BtaF) in the adhesive properties of the bacterial surface. BtaF conferred the bacteria that carried it a promiscuous adhesiveness to various ECM components and the ability to attach to an abiotic surface. Furthermore, BtaF was found to participate in bacterial adhesion to epithelial cells and was required for full virulence in mice. Similar to BmaC and BtaE, the BtaF adhesin was expressed in a small subpopulation of bacteria, and in all cases, it was detected at the new pole generated after cell division. Interestingly, BtaF was also implicated in the resistance of B. suis to porcine serum. Our findings emphasize the impact of TAs in the Brucella lifecycle.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion/physiology , Brucella suis/physiology , Brucella suis/pathogenicity , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/immunology , Animals , Brucellosis/immunology , Brucellosis/metabolism , Cell Line , Extracellular Matrix/metabolism , Humans , Male , Mice , Multigene Family , Protein Multimerization , Protein Transport , Swine , Virulence
8.
BMC Microbiol ; 13: 199, 2013 Sep 04.
Article in English | MEDLINE | ID: mdl-24007556

ABSTRACT

BACKGROUND: During the infection process, bacteria are confronted with various stress factors including nutrient starvation. In an in vitro model, adaptation strategies of nutrient-starved brucellae, which are facultative intracellular pathogens capable of long-term persistence, were determined. RESULTS: Long-term nutrient starvation in a medium devoid of carbon and nitrogen sources resulted in a rapid decline in viability of Brucella suis during the first three weeks, followed by stabilization of the number of viable bacteria for a period of at least three weeks thereafter. A 2D-Difference Gel Electrophoresis (DIGE) approach allowed the characterization of the bacterial proteome under these conditions. A total of 30 proteins showing altered concentrations in comparison with bacteria grown to early stationary phase in rich medium were identified. More than half of the 27 significantly regulated proteins were involved in bacterial metabolism with a marked reduction of the concentrations of enzymes participating in amino acid and nucleic acid biosynthesis. A total of 70% of the significantly regulated proteins showed an increased expression, including proteins involved in the adaptation to harsh conditions, in regulation, and in transport. CONCLUSIONS: The adaptive response of Brucella suis most likely contributes to the long-term survival of the pathogen under starvation conditions, and may play a key role in persistence.


Subject(s)
Adaptation, Physiological , Bacterial Proteins/analysis , Brucella suis/physiology , Proteome/analysis , Brucella suis/chemistry , Brucella suis/metabolism , Carbon/metabolism , Culture Media/chemistry , Electrophoresis, Gel, Two-Dimensional , Microbial Viability , Nitrogen/metabolism
9.
BMC Genomics ; 14: 459, 2013 Jul 08.
Article in English | MEDLINE | ID: mdl-23834488

ABSTRACT

BACKGROUND: In the intracellular pathogen Brucella spp., the activation of the stringent response, a global regulatory network providing rapid adaptation to growth-affecting stress conditions such as nutrient deficiency, is essential for replication in the host. A single, bi-functional enzyme Rsh catalyzes synthesis and hydrolysis of the alarmone (p)ppGpp, responsible for differential gene expression under stringent conditions. RESULTS: cDNA microarray analysis allowed characterization of the transcriptional profiles of the B. suis 1330 wild-type and Δrsh mutant in a minimal medium, partially mimicking the nutrient-poor intramacrophagic environment. A total of 379 genes (11.6% of the genome) were differentially expressed in a rsh-dependent manner, of which 198 were up-, and 181 were down-regulated. The pleiotropic character of the response was confirmed, as the genes encoded an important number of transcriptional regulators, cell envelope proteins, stress factors, transport systems, and energy metabolism proteins. Virulence genes such as narG and sodC, respectively encoding respiratory nitrate reductase and superoxide dismutase, were under the positive control of (p)ppGpp, as well as expression of the cbb3-type cytochrome c oxidase, essential for chronic murine infection. Methionine was the only amino acid whose biosynthesis was absolutely dependent on stringent response in B. suis. CONCLUSIONS: The study illustrated the complexity of the processes involved in adaptation to nutrient starvation, and contributed to a better understanding of the correlation between stringent response and Brucella virulence. Most interestingly, it clearly indicated (p)ppGpp-dependent cross-talk between at least three stress responses playing a central role in Brucella adaptation to the host: nutrient, oxidative, and low-oxygen stress.


Subject(s)
Adaptation, Physiological/genetics , Bacterial Proteins/metabolism , Brucella suis/genetics , Brucella suis/physiology , Gene Expression Profiling , Stress, Physiological/genetics , Animals , Brucella suis/enzymology , Brucella suis/metabolism , Electron Transport Complex IV/genetics , Macrophages/cytology , Methionine/biosynthesis , Mice , Mutation , Nitrate Reductase/genetics , Oligonucleotide Array Sequence Analysis , Superoxide Dismutase/genetics , Up-Regulation , Vacuoles/microbiology
10.
Cell Microbiol ; 14(6): 965-82, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22321605

ABSTRACT

Brucella is an intracellular pathogen responsible of a zoonotic disease called brucellosis. Brucella survives and proliferates within several types of phagocytic and non-phagocytic cells. Like in other pathogens, adhesion of brucellae to host surfaces was proposed to be an important step in the infection process. Indeed, Brucella has the capacity to bind to culture human cells and key components of the extracellular matrix, such as fibronectin. However, little is known about the molecular bases of Brucella adherence. In an attempt to identify bacterial genes encoding adhesins, a phage display library of Brucella suis was panned against fibronectin. Three fibronectin-binding proteins of B. suis were identified using this approach. One of the candidates, designated BmaC was a very large protein of 340 kDa that is predicted to belong to the type I (monomeric) autotransporter family. Microscopy studies showed that BmaC is located at one pole on the bacterial surface. The phage displaying the fibronectin-binding peptide of BmaC inhibited the attachment of brucellae to both, HeLa cells and immobilized fibronectin in vitro. In addition, a bmaC deletion mutant was impaired in the ability of B. suis to attach to immobilized fibronectin and to the surface of HeLa and A549 cells and was out-competed by the wild-type strain in co-infection experiments. Finally, anti-fibronectin or anti-BmaC antibodies significantly inhibited the binding of wild-type bacteria to HeLa cells. Our results highlight the role of a novel monomeric autotransporter protein in the adhesion of B. suis to the extracellular matrix and non-phagocytic cells via fibronectin binding.


Subject(s)
Adhesins, Bacterial/physiology , Bacterial Adhesion , Brucella suis/physiology , Host-Pathogen Interactions , Membrane Transport Proteins/physiology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Animals , Brucella suis/growth & development , Brucella suis/metabolism , Fibronectins/chemistry , Fibronectins/metabolism , Gene Knockout Techniques , HeLa Cells , Humans , Immobilized Proteins/chemistry , Macrophages/microbiology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Microbial Viability , Peptide Library , Protein Structure, Tertiary , Sequence Analysis, DNA
11.
Res Vet Sci ; 93(2): 565-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-21968103

ABSTRACT

A new selective medium, named LNIV-M, has been developed for isolation of Brucella suis. In this work, we evaluated the growth of B. suis reference and field strains from domestic pigs in different basal media and the susceptibility to different antibiotics contained in the currently used Farrell's and modified Thayer-Martin media. We also determined the efficacy of LNIV-M and its diagnostic performance for isolating B. suis from wild boar tissue samples. A total of 1649 samples from 918 hunter-harvested wild boars were cultured in LNIV-M, Farrell's and modified Thayer-Martin media. One hundred and thirty-nine (8.4%) samples from 63 (6.9%) animals resulted in a positive culture. LNIV-M detected 93.6% and 62.6% of positive animals and samples, respectively, while Farrell's and modified Thayer-Martin media detected, respectively, 92.1% and 79.4% of positive animals and 58.3% and 59.7% of samples. These results confirm the adequate diagnostic performance of LNIV-M in the isolation of B. suis.


Subject(s)
Bacteriological Techniques/veterinary , Brucella suis/physiology , Brucellosis/veterinary , Culture Media/chemistry , Swine Diseases/diagnosis , Animals , Anti-Bacterial Agents/pharmacology , Bacteriological Techniques/standards , Brucella suis/drug effects , Brucella suis/isolation & purification , Brucellosis/diagnosis , Brucellosis/microbiology , Culture Media/standards , Drug Resistance, Bacterial , Microbial Sensitivity Tests , Sus scrofa , Swine , Swine Diseases/microbiology
12.
Infect Immun ; 79(6): 2460-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21464087

ABSTRACT

Brucella spp. are intracellular bacteria that cause an infectious disease called brucellosis in humans and many domestic and wildlife animals. B. suis primarily infects pigs and is pathogenic to humans. The macrophage-Brucella interaction is critical for the establishment of a chronic Brucella infection. Our studies showed that smooth virulent B. suis strain 1330 (S1330) prevented programmed cell death of infected macrophages and rough attenuated B. suis strain VTRS1 (a vaccine candidate) induced strong macrophage cell death. To further investigate the mechanism of VTRS1-induced macrophage cell death, microarrays were used to analyze temporal transcriptional responses of murine macrophage-like J774.A1 cells infected with S1330 or VTRS1. In total 17,685 probe sets were significantly regulated based on the effects of strain, time and their interactions. A miniTUBA dynamic Bayesian network analysis predicted that VTRS1-induced macrophage cell death was mediated by a proinflammatory gene (the tumor necrosis factor alpha [TNF-α] gene), an NF-κB pathway gene (the IκB-α gene), the caspase-2 gene, and several other genes. VTRS1 induced significantly higher levels of transcription of 40 proinflammatory genes than S1330. A Mann-Whitney U test confirmed the proinflammatory response in VTRS1-infected macrophages. Increased production of TNF-α and interleukin 1ß (IL-1ß) were also detected in the supernatants in VTRS1-infected macrophage cell culture. Hyperphosphorylation of IκB-α was observed in macrophages infected with VTRS1 but not S1330. The important roles of TNF-α and IκB-α in VTRS1-induced macrophage cell death were further confirmed by individual inhibition studies. VTRS1-induced macrophage cell death was significantly inhibited by a caspase-2 inhibitor but not a caspase-1 inhibitor. The role of caspase-2 in regulating the programmed cell death of VTRS1-infected macrophages was confirmed in another study using caspase-2-knockout mice. In summary, VTRS1 induces a proinflammatory, caspase-2- and NF-κB-mediated macrophage cell death. This unique cell death differs from apoptosis, which is not proinflammatory. It is also different from classical pyroptosis, which is caspase-1 mediated.


Subject(s)
Brucella suis/physiology , Brucellosis/microbiology , Caspase 2/physiology , Macrophages/microbiology , Animals , Brucella suis/immunology , Brucellosis/immunology , Cell Death , Gene Expression Profiling , Gene Expression Regulation, Bacterial/physiology , Interleukin-1beta/physiology , Macrophages/immunology , Mice , Oligonucleotide Array Sequence Analysis , Phosphorylation , Tumor Necrosis Factor-alpha/physiology
13.
Org Biomol Chem ; 9(10): 3681-90, 2011 May 21.
Article in English | MEDLINE | ID: mdl-21461427

ABSTRACT

In the facultative intracellular pathogen Brucella suis, histidinol dehydrogenase (HDH) activity, catalyzing the last step in histidine biosynthesis, is essential for intramacrophagic replication. The inhibition of this virulence factor by substituted benzylic ketones was a proof of concept that disarming bacteria leads to inhibition of intracellular bacterial growth in macrophage infection. This work describes the design, synthesis and evaluation of 19 new potential HDH inhibitors, using a combination of classical approaches and docking studies. The IC(50)-values of these inhibitors on HDH activity were in the nanomolar range, and several of them showed a 70-100% inhibition of Brucella growth in minimal medium. One selected compound yielded a strong inhibitory effect on intracellular replication of B. suis in human macrophages at concentrations as low as 5 µM, with an overall survival of intramacrophagic bacteria reduced by a factor 10(3). Docking studies with two inhibitors showed a good fitting in the catalytic pocket and also interaction with the second lipophilic pocket binding the cofactor NAD(+). Experimental data confirmed competition between inhibitors and NAD(+) at this site. Hence, these inhibitors can be considered as promising tools in the development of novel anti-virulence drugs.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Brucella suis/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Models, Molecular , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Binding, Competitive , Brucella suis/enzymology , Brucella suis/pathogenicity , Brucella suis/physiology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Ketones/chemistry , Macrophages/drug effects , Macrophages/microbiology , Molecular Sequence Data , NAD/metabolism , Protein Conformation , Substrate Specificity , Virus Replication/drug effects
14.
J Genet Genomics ; 37(11): 725-36, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21115167

ABSTRACT

Brucella suis is responsible for swine brucellosis worldwide. Of the five different B. suis biovars (bv.), bv. 2 appears restricted to Europe where it is frequently isolated from wild boar and hares, can infect pigs and can cause human brucellosis. In this study, the differential gene expression profile was characterized in spleens of Eurasian wild boar naturally infected with B. suis bv. 2. Of the 20,201 genes analyzed in the microarray, 633 and 1,373 were significantly (fold change > 1.8; P < 0.01) upregulated and downregulated, respectively, in infected wild boar. The analysis was focused on genes that were over represented after conditional test for biological process gene ontology. Upregulated genes suggested that B. suis bv. 2 infection induced cell maturation, migration and/or proliferation in infected animals. The genes downregulated in infected wild boar impaired the activity of several important cellular metabolic pathways such as metabolism, cytoskeleton organization and biogenesis, immune response and lysosomal function and vesicle-mediated transport. In addition, the response to stress, sperm fertility, muscle development and apoptosis seemed to be also impaired in infected animals. These results suggested that B. suis bv. 2 may use strategies similar to other smooth brucellae to facilitate intracellular multiplication and the development of chronic infections. To our knowledge, this is the first report of the analysis of gene expression profile in hosts infected with B. suis bv. 2, which is important to understand the molecular mechanisms at the host-pathogen interface in the main reservoir species with possible implications in the zoonotic cycle of the pathogen.


Subject(s)
Brucella suis/physiology , Brucellosis/veterinary , Disease Reservoirs/microbiology , Gene Expression Profiling , Spleen/microbiology , Swine Diseases/genetics , Swine Diseases/microbiology , Animals , Animals, Wild/genetics , Animals, Wild/metabolism , Animals, Wild/microbiology , Brucella suis/classification , Brucella suis/isolation & purification , Brucellosis/genetics , Brucellosis/metabolism , Brucellosis/microbiology , Gene Expression Regulation , Male , Molecular Sequence Data , Spleen/metabolism , Sus scrofa/microbiology , Swine , Swine Diseases/metabolism
15.
J Bacteriol ; 191(8): 2530-40, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19201794

ABSTRACT

The RND-type efflux pumps are responsible for the multidrug resistance phenotype observed in many clinically relevant species. Also, RND pumps have been implicated in physiological processes, with roles in the virulence mechanisms of several pathogenic bacteria. We have previously shown that the BepC outer membrane factor of Brucella suis is involved in the efflux of diverse drugs, probably as part of a tripartite complex with an inner membrane translocase. In the present work, we characterize two membrane fusion protein-RND translocases of B. suis encoded by the bepDE and bepFG loci. MIC assays showed that the B. suis DeltabepE mutant was more sensitive to deoxycholate (DOC), ethidium bromide, and crystal violet. Furthermore, multicopy bepDE increased resistance to DOC and crystal violet and also to other drugs, including ampicillin, norfloxacin, ciprofloxacin, tetracycline, and doxycycline. In contrast to the DeltabepE mutant, the resistance profile of B. suis remained unaltered when the other RND gene (bepG) was deleted. However, the DeltabepE DeltabepG double mutant showed a more severe phenotype than the DeltabepE mutant, indicating that BepFG also contributes to drug resistance. An open reading frame (bepR) coding for a putative regulatory protein of the TetR family was found upstream of the bepDE locus. BepR strongly repressed the activity of the bepDE promoter, but DOC released the repression mediated by BepR. A clear induction of the bepFG promoter activity was observed only in the BepDE-defective mutant, indicating a regulatory interplay between the two RND efflux pumps. Although only the BepFG-defective mutant showed a moderate attenuation in model cells, the activities of both bepDE and bepFG promoters were induced in the intracellular environment of HeLa cells. Our results show that B. suis harbors two functional RND efflux pumps that may contribute to virulence.


Subject(s)
Bacterial Proteins/metabolism , Brucella suis/drug effects , Drug Resistance, Bacterial , Membrane Transport Proteins/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Brucella suis/pathogenicity , Brucella suis/physiology , Deoxycholic Acid/pharmacology , Epithelial Cells/microbiology , Ethidium/pharmacology , Gene Deletion , Gene Dosage , Gene Expression Regulation, Bacterial , Gentian Violet/pharmacology , HeLa Cells , Humans , Membrane Transport Proteins/genetics , Microbial Sensitivity Tests , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Alignment , Virulence
16.
Infect Immun ; 73(10): 6229-36, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16177294

ABSTRACT

Brucella spp. are stealthy bacteria that enter host cells without major perturbation. The molecular mechanism involved is still poorly understood, although numerous studies have been published on this subject. Recently, it was reported that Brucella abortus utilizes cellular prion protein (PrP(C)) to enter the cells and to reach its replicative niche. The molecular mechanisms involved were not clearly defined, prompting us to analyze this process using blocking antibodies against PrP(C). However, the behavior of Brucella during cellular infection under these conditions was not modified. In a next step, the behavior of Brucella in macrophages lacking the prion gene and the infection of mice knocked out for the prion gene were studied. We observed no difference from results obtained with the wild-type control. Although some contacts between PrP(C) and Brucella were observed on the surface of the cells by using confocal microscopy, we could not show that Brucella specifically bound recombinant PrP(C). Therefore, we concluded from our results that prion protein (PrP(C)) was not involved in Brucella infection.


Subject(s)
Brucella suis/physiology , Brucellosis/etiology , Macrophages/microbiology , PrPC Proteins/physiology , Animals , Antibodies/pharmacology , Brucella suis/chemistry , Brucellosis/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Chaperonin 60/analysis , Chaperonin 60/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Phagosomes/metabolism , PrPC Proteins/antagonists & inhibitors , PrPC Proteins/genetics
17.
Vet Microbiol ; 109(1-2): 95-104, 2005 Aug 10.
Article in English | MEDLINE | ID: mdl-15970403

ABSTRACT

In Gram-negative bacteria, autotransporters are secreted proteins able to translocate themselves through the inner- and outer-membranes to the cell surface or to the extracellular environment. The influence of the putative outer membrane autotransporter (OmaA) protein to the persistence of Brucella suis was investigated. Sequence analyses revealed that the OmaA protein of B. suis strain 1330 consists of a signal peptide, a passenger alpha-domain, and a transporter beta-domain, which are the characteristic components of an autotransporter protein. The transporter beta-domain consists of 14 individual amphipathic beta-strands, and a 46-amino acid long alpha-helix lies upstream of the transporter domain, indicating that the B. suis OmaA is a type-I classical autotransporter. BLAST search and phylogenetic analyses revealed that the B. suis OmaA protein shares more similarities with adhesin autotransporter proteins than with protease autotransporter proteins of other bacteria. An OmaA-deficient strain (1330DeltaomaA) was generated by disrupting the DNA region encoding the passenger alpha-domain of the OmaA protein of B. suis wild type strain 1330. The omaA gene encoding the full-length OmaA protein was cloned and used to complement the OmaA-deficient strain. The OmaA-deficient strain did not differ from the wild type strain in terms of persistence in J774 macrophage cell line 24 and 48 h after inoculation, or clearance from the spleens of BALB/c mice at 1 week after intraperitoneal inoculation. These observations suggest that the function of the OmaA protein is dispensable during the acute phase of B. suis infection. However, the OmaA-deficient strain was cleared from the spleens of BALB/c mice faster than the wild type strain between the third and the ninth week after intraperitoneal inoculation, indicating that the OmaA may be important during the chronic phase of B. suis infection. Relative to the BALB/c mice injected with saline, those vaccinated with the OmaA-deficient strain exhibited 3.0-3.9log10 colony forming units protection against a challenge with B. suis strain 1330. This study is the first report correlating an autotransporter protein deficiency with persistence of B. suis in vitro and in vivo.


Subject(s)
Bacterial Outer Membrane Proteins/physiology , Brucella suis/physiology , Brucellosis/microbiology , Carrier Proteins/physiology , Animals , Bacterial Outer Membrane Proteins/genetics , Brucella suis/genetics , Brucella suis/growth & development , Carrier Proteins/genetics , Colony Count, Microbial , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Female , Genetic Complementation Test , Macrophages , Mice , Mice, Inbred BALB C , Mutagenesis, Insertional , Phylogeny , Recombinant Proteins , Sequence Analysis, DNA , Spleen/microbiology
18.
Proc Natl Acad Sci U S A ; 102(12): 4596-601, 2005 Mar 22.
Article in English | MEDLINE | ID: mdl-15764702

ABSTRACT

Type IV secretion systems (T4SSs) are commonly used secretion machineries in Gram-negative bacteria. They are used in the infection of human, animal, or plant cells and the propagation of antibiotic resistance. The T4SS apparatus spans both membranes of the bacterium and generally is composed of 12 proteins, named VirB1-11 and VirD4 after proteins of the canonical Agrobacterium tumefaciens T4SS. The periplasmic core complex of VirB8/VirB10 structurally and functionally links the cytoplasmic NTPases of the system with its outer membrane and pilus components. Here we present crystal structures of VirB8 of Brucella suis, the causative agent of brucellosis, and ComB10, a VirB10 homolog of Helicobacter pylori, the causative agent of gastric ulcers. The structures of VirB8 and ComB10 resemble known folds, albeit with novel secondary-structure modifications unique to and conserved within their respective families. Both proteins crystallized as dimers, providing detailed predictions about their self associations. These structures make a substantial contribution to the repertoire of T4SS component structures and will serve as springboards for future functional and protein-protein interaction studies by using knowledge-based site-directed and deletion mutagenesis.


Subject(s)
Bacterial Proteins/chemistry , Brucella suis/chemistry , Helicobacter pylori/chemistry , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Brucella suis/genetics , Brucella suis/pathogenicity , Brucella suis/physiology , Cloning, Molecular , Conserved Sequence , Crystallography, X-Ray , Dimerization , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Helicobacter pylori/physiology , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Homology, Amino Acid , Static Electricity
19.
Vet Microbiol ; 90(1-4): 299-309, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12414150

ABSTRACT

Phagocytes have developed various antimicrobial defense mechanisms to eliminate pathogens. They comprise the oxidative burst, acidification of phagosomes, or fusion of phagosomes with lysosomes. Facultative intracellular bacteria, in return, have developed strategies counteracting the host cell defense, resulting in intramacrophagic survival. Until lately, only very little was known about the phagosomal compartment containing Brucella spp., the environmental conditions the bacteria encounter, and the pathogen's stress response. Recently, we have determined that the phagosomes acidify rapidly to a pH of 4.0-4.5 following infection, but this early acidification is crucial for intracellular replication as neutralization results in bacterial elimination. A vacuolar proton-ATPase is responsible for this phenomenon that is not linked to phagosome-lysosome fusion. On the contrary, in vitro reconstitution assays revealed association only between phagosomes containing killed B. suis and lysosomes, describing the absence of phagolysosome fusion due to specific recognition inhibition for live bacteria. Further evidence for the necessity of an intact, acidic phagosome as a predominant niche of brucellae in macrophages was obtained with a strain of B. suis secreting listeriolysin. It partially disrupts the phagosomal membranes and fails to multiply intracellularly. How does B. suis adapt to this environment? We have identified and studied a series of genes that are involved in this process of adaptation. The bacterial heat shock protein and chaperone DnaK is induced in phagocytes and it is essential for intracellular multiplication. A low-level, constitutive expression of dnaK following promoter exchange does not restore intramacrophagic survival. Another chaperone and heat shock protein, ClpB, belonging to the family of ClpATPases, is important for the resistance of B. suis to several in vitro stresses, but does not contribute to intramacrophagic survival of the pathogen. Additional bacterial genes specifically induced within the phagocyte were identified by an intramacrophagic screen of random promoter fusions to the reporter gene gfp. A large majority of these genes are encoding proteins involved in transport of nutrients (sugars, amino acids), or cofactors, such as nickel. Analysis of the intracellular gene activation reveals that low oxygen tension is encountered by B. suis. Altogether, these results suggest three major stress conditions encountered by brucellae in the phagosome: acid stress, starvation and low oxygen tension.


Subject(s)
Brucella suis/physiology , Brucella suis/pathogenicity , Macrophages/microbiology , Animals , Brucella suis/ultrastructure , Humans , Hydrogen-Ion Concentration , Lysosomes/physiology , Operon , Phagosomes/physiology
20.
Proc Natl Acad Sci U S A ; 99(24): 15711-6, 2002 Nov 26.
Article in English | MEDLINE | ID: mdl-12438693

ABSTRACT

The pathogen Brucella suis resides and multiplies within a phagocytic vacuole of its host cell, the macrophage. The resulting complex relationship has been investigated by the analysis of the set of genes required for virulence, which we call intramacrophagic virulome. Ten thousand two hundred and seventy-two miniTn5 mutants of B. suis constitutively expressing gfp were screened by fluorescence microscopy for lack of intracellular multiplication in human macrophages. One hundred thirty-one such mutants affected in 59 different genes could be isolated, and a function was ascribed to 53 of them. We identified genes involved in (i) global adaptation to the intracellular environment, (ii) amino acid, and (iii) nucleotide synthesis, (iv) sugar metabolism, (v) oxidoreduction, (vi) nitrogen metabolism, (vii) regulation, (viii) disulphide bond formation, and (ix) lipopolysaccharide biosynthesis. Results led to the conclusion that the replicative compartment of B. suis is poor in nutrients and characterized by low oxygen tension, and that nitrate may be used for anaerobic respiration. Intramacrophagic virulome analysis hence allowed the description of the nature of the replicative vacuole of the pathogen in the macrophage and extended our understanding of the niche in which B. suis resides. We propose calling this specific compartment "brucellosome."


Subject(s)
Bacterial Proteins/genetics , Brucella suis/pathogenicity , Genes, Bacterial , Macrophages/microbiology , Adaptation, Physiological , Amino Acids/biosynthesis , Anaerobiosis , Bacterial Proteins/biosynthesis , Brucella suis/genetics , Brucella suis/physiology , Carbohydrate Metabolism , Cell Division , Electron Transport , Genes, Reporter , Green Fluorescent Proteins , Lipid Metabolism , Lipopolysaccharides/metabolism , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Microscopy, Fluorescence , Mutagenesis, Insertional , Nitrogen/metabolism , Nucleotides/biosynthesis , Vacuoles/microbiology , Virulence/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...